EP3655379B1 - Verfahren zur herstellung von verbindungen mit bis-tetrahydroisochinolin - Google Patents

Verfahren zur herstellung von verbindungen mit bis-tetrahydroisochinolin Download PDF

Info

Publication number
EP3655379B1
EP3655379B1 EP18834872.6A EP18834872A EP3655379B1 EP 3655379 B1 EP3655379 B1 EP 3655379B1 EP 18834872 A EP18834872 A EP 18834872A EP 3655379 B1 EP3655379 B1 EP 3655379B1
Authority
EP
European Patent Office
Prior art keywords
alkyl
compound
iridium
alkoxy
bis
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Active
Application number
EP18834872.6A
Other languages
English (en)
French (fr)
Other versions
EP3655379A4 (de
EP3655379A1 (de
Inventor
Brian M. Stoltz
Eric R. WELIN
Scott C. Virgil
Pamela TADROSS
Gerit Maria POTOTSCHNIG
Aurapat NGAMNITHIPORN (Fa)
Kenji Negoro
Guillaume LAPOINTE
Max KLATTE
Christopher HALEY
Christian Gruenanger
Emil GLIBSTRUP
Christopher GILMORE
Kevin McCormack ALLAN
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
California Institute of Technology CalTech
Original Assignee
California Institute of Technology CalTech
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by California Institute of Technology CalTech filed Critical California Institute of Technology CalTech
Publication of EP3655379A1 publication Critical patent/EP3655379A1/de
Publication of EP3655379A4 publication Critical patent/EP3655379A4/de
Application granted granted Critical
Publication of EP3655379B1 publication Critical patent/EP3655379B1/de
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/12Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains three hetero rings
    • C07D471/18Bridged systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/04Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings directly linked by a ring-member-to-ring-member bond

Definitions

  • bis-THIQ bis-tetrahydroisoquinoline
  • Jorumycin is considered the minimum pharmacophore of this natural product family, possessing a pentacyclic core, polyoxygenated termini, and carbinolamine functionality that lend these natural products their marked biological activity.
  • electrophilic aromatic chemistry such as the Pictet-Spengler reaction, which has limited the synthesis of non-natural analogs to highly electron-rich species that facilitate this reactivity.
  • the present disclosure relates in part to the synthesis of jorumycin and structurally-related compounds, e.g., using a concise and convergent cross-coupling/enantioselective isoquinoline hydrogenation strategy, providing bis-THIQ compounds, including electron-deficient bis-THIQ variants.
  • the present disclosure provides a method for preparing a compound of Formula (I): comprising contacting a compound of Formula (II): with a transition metal catalyst (preferably a chiral transition metal catalyst) under hydrogenation conditions, wherein, as valence and stability permit:
  • preparing the compound of Formula (II): comprises combining a compound of Formula (III): a compound of Formula (IV): and a transition metal catalyst under cross-coupling conditions, wherein, as valence and stability permit:
  • bis-THIQ bis-tetrahydroisoquinoline
  • Et 743 Yondelis ® , trabectidin
  • 2 is available from nature, isolation of one gram of material would require in excess of one ton of biological material. 5 For this reason, the successful application of 2 as an anti-tumor agent has necessitated its large-scale chemical synthesis, a daunting task for a molecule of such immense complexity.
  • Jorumycin Since its isolation in 2000, 15 jorumycin been the target of a number of synthetic endeavors, including four total syntheses 16-19 and two semi-syntheses. 20,21 Similar to the strategies discussed above, EAS-based reactions have been heavily utilized in these studies. Jorumycin possesses all of the defining features of the bis-THIQ natural products-the penta-cyclic carbon skeleton, the polyoxygenated ring termini, and the central carbinolamine-that together provide the marked bio-logical activity of this natural product family.
  • 2-4 Jorumycin exhibits an IC 50 of 0.24 nM vs. A549 lung cancer, 0.49 nM vs. DU145 prostate cancer, and 0.57 nM vs. HCT116 colon cancer, 15,19,20 thus offering immense therapeutic potential.
  • the present disclosure provides a method for preparing a compound of Formula (I): comprising contacting a compound of Formula (II): with a transition metal catalyst (preferably a chiral transition metal catalyst) under
  • each instance of R 1 , R 2 , R 3 , R 4 , and R 5 is independently unsubstituted alkyl or alkyl substituted with one or more substituents selected from hydroxy, alkoxy, acyloxy, amino, and thio.
  • the chiral ligand comprises a diphosphine ligand, preferably a ferrocenyl diphosphine ligand.
  • the diphosphine ligand is selected from S -(CF 3 )- t -BuPHOX, S,S -Et-FerroTANE, S,R p - xyliphos, or S,R p -BTFM-xyliphos, R -(CF 3 )- t -BuPHOX, R,R -Et-FerroTANE, R,S p - xyliphos, and R,S p -BTFM-xyliphos.
  • the chiral ligand is S,R p -BTFM-xyliphos or R,S p -BTFM-xyliphos.
  • the transition metal catalyst is a chiral transition metal catalyst and is used in an amount from about 0.1 mol% to about 100 mol% relative to the compound of formula (II) or (VII). In other embodiments, the transition metal catalyst is a chiral transition metal catalyst and is used in an amount from about 5 mol% to about 30 mol% relative to the compound of formula (II) or (VII). In certain embodiments, the iridium catalyst is used in an amount of about 20 mol% relative to the compound of formula (II) or (VII).
  • R 13 and R 14 are each independently unsubstituted alkyl or alkyl substituted with one or more substituents selected from hydroxy, alkoxy, acyloxy, amino and thio. In certain embodiments, R 14 is hydroxyalkyl.
  • the transition metal catalyst comprises a nickel (e.g., Ni(COD) 2 ), palladium, or platinum catalyst. In certain embodiments, the transition metal catalyst comprises a palladium catalyst. In some embodiments, the palladium catalyst is selected from Pd/C, Pd 2 (DBA) 3 , Pd(PPh 3 ) 4 , Pd(OC(O)R C ) 2 , Pd(OAc) 2 , PdCl 2 , Pd(PhCN) 2 Cl 2 , Pd(CH 3 CN) 2 Cl 2 , PdBr 2 , Pd(acac) 2 , [Pd(allyl)Cl] 2 , Pd(TFA) 2 , Pd 2 (pmdba) 3 , Pd(P(t-Bu) 2 Me) 2 , and pre-formed Pd(II)-ligand complexes; wherein R C is optionally substituted alkyl, alkenyl, alkynyl, ary
  • the palladium catalyst is Pd(P(t-Bu) 2 Me) 2 .
  • the transition metal catalyst is used in an amount from about 0.5 mol% to about 50 mol% relative to the compound of formula (III) or (IV).
  • R 1 is hydrogen. In other embodiments, R 1 is unsubstituted alkyl or alkyl substituted with one or more substituents selected from hydroxy, alkoxy, acyloxy, amino, and thio. In some embodiments, R 2 is selected from H, halo and hydroxy. In some embodiments, R 3 is alkyl. In other embodiments, R 5 is hydroxy or alkoxy. In some embodiments, R 6 is unsubstituted alkyl or alkyl substituted with one or more substituents selected from hydroxy, alkoxy, acyloxy, amino and thio. In some embodiments, R 6 is hydroxyalkyl or acyloxyalkyl, preferably CH 2 OH.
  • R 7 is unsubstituted alkyl or alkyl substituted with one or more substituents selected from hydroxy, alkoxy, acyloxy, amino, and thio. In other embodiments, R 7 is H, halo or hydroxy. In some embodiments, R 8 is alkoxy. In some embodiments, R 9 is alkyl. In certain embodiments, R 10 is hydroxy or alkoxy. In some embodiments, R 11 is H.
  • each instance of R 2 , R 3 , R 4 , R 5 , R 8 , R 9 , R 10 , and R 11 is independently unsubstituted alkyl or alkyl substituted with one or more substituents selected from hydroxy, alkoxy, acyloxy, amino, and thio.
  • the compound may have the formula (VA):
  • the compound may have the formula (VB):
  • the compound may have the Formula (VI):
  • the compound may have the Formula (VIA):
  • the compound may have the Formula (VIB):
  • R 1 , R 6 , and R 7 may each be unsubstituted alkyl or alkyl substituted with one or more substituents selected from hydroxy, alkoxy, acyloxy, amino, and thio.
  • Each instance of R 2 , R 3 , R 4 , R 5 , R 8 , R 9 , R 10 , and R 11 may independently be unsubstituted alkyl or alkyl substituted with one or more substituents selected from hydroxy, alkoxy, acyloxy, amino, and thio.
  • R 15 may be H or alkyl.
  • R 15 may be unsubstituted alkyl or alkyl substituted with one or more substituents selected from hydroxy, alkoxy, acyloxy, amino, and thio.
  • R 15 is methyl.
  • X may be unsubstituted alkyl or alkyl substituted with one or more substituents selected from hydroxy, alkoxy, acyloxy, amino, and thio.
  • Each R 3 , R 4 , R 8 , or R 9 may be independently carbonyl, halogen, nitro, cyano, carboxyl, sulfate, amino, alkoxy, alkylamino, ester, sulfonate, sulfone, sulfoxide, acyl, haloalkyl or acyloxy.
  • Each R 3 , R 4 , R 8 , or R 9 may be independently carbonyl, halogen, nitro, cyano, carboxyl, ester, acyl, haloalkyl or acyloxy.
  • the compound may be a prodrug, e.g., wherein a hydroxyl in the parent compound is presented as an ester or a carbonate, a carboxylic acid present in the parent compound is presented as an ester, or an amino group is presented as an amide.
  • the prodrug may be metabolized to the active parent compound in vivo (e.g., the ester is hydrolyzed to the corresponding hydroxyl or carboxylic acid).
  • the compounds described may be racemic.
  • Thecompounds may be enriched in one enantiomer.
  • a compound may have greater than 30% ee, 40% ee, 50% ee, 60% ee, 70% ee, 80% ee, 90% ee, 95% ee, 96% ee, 97% ee, 98% ee, 99% or greater ee.
  • the compounds have more than one stereocenter.
  • the compounds may be enriched in one or more diastereomers.
  • a compound may have greater than 30% de, 40% de, 50% de, 60% de, 70% de, 80% de, 90% de, 95% de, 96% de, 97% de, 98% de, 99% or greater de.
  • the compounds may have substantially one isomeric configuration at one or more stereogenic centers, and may have multiple isomeric configurations at the remaining stereogenic centers.
  • a therapeutic preparation of the compound may be enriched to provide predominantly one enantiomer of a compound.
  • An enantiomerically enriched mixture may comprise, for example, at least 60 mol percent of one enantiomer, or more preferably at least 75, 90, 95, or even 99 mol percent.
  • the compound enriched in one enantiomer may be substantially free of the other enantiomer, wherein substantially free means that the substance in question makes up less than 10%, or less than 5%, or less than 4%, or less than 3%, or less than 2%, or less than 1% as compared to the amount of the other enantiomer, e.g., in the composition or compound mixture.
  • a composition or compound mixture contains 98 grams of a first enantiomer and 2 grams of a second enantiomer, it would be said to contain 98 mol percent of the first enantiomer and only 2% of the second enantiomer.
  • a therapeutic preparation may be enriched to provide predominantly one diastereomer of the compound.
  • a diastereomerically enriched mixture may comprise, for example, at least 60 mol percent of one diastereomer, or more preferably at least 75, 90, 95, or even 99 mol percent.
  • the bis-THIQcompounds prepared by the method of the invention exhibit an improved pharmacokinetic profile relative to existing bis-THIQs.
  • the bis-THIQ compounds prepared by the method of the invention exhibit improved bioavailability relative to existing bis-THIQs.
  • Preferred transition metal catalysts of the invention are complexes of iridium.
  • the transition metal catalyst is an iridium catalyst.
  • the iridium catalyst is prepared by combining an iridium source and a chiral ligand. In preferred embodiments the iridium catalyst is prepared by combining an iridium source and a chiral ligand.
  • Exemplary iridium sources that may be used in the methods of the invention include, but are not limited to, (acetylacetonato)(1,5-cyclooctadiene)iridium(I), (acetylacetonato)(1,5-cyclooctadiene)iridium(I), (acetylacetonato)dicarbonyliridium(I), bis[1,2-bis(diphenylphosphino)ethane]carbonyl chloroiridium(I), bis(1,5-cyclooctadiene)diiridium(I) dichloride, bis(1,5-cyclooctadiene)iridium(I) tetrafluoroborate, bis(cyclooctadiene)iridium(I) tetrakis(3,5-bis(trifluoromethyl)phenyl)borate, chlorobis(cyclooctene)iridium(I)dimer, (1,5
  • Catalyst loading may be expressed as a percentage that is calculated by dividing the moles of catalyst complex by the moles of the substrate present in a given reaction.
  • Catalyst loading is alternatively expressed as a percentage that is calculated by dividing the moles of total transition metal (for example, iridium) by the moles of the substrate present in a given reaction.
  • the transition metal catalyst is present under the conditions of the reaction from an amount of about 0.01 mol% to about 30 mol% total iridium relative to the substrate, such as the compound of formula (II) or (VII).
  • the catalyst loading is from about 0.05 mol% to about 25 mol% total iridium relative to the substrate.
  • the catalyst loading is from about 0.1 mol% to about 25 mol%, about 1 mol% to about 25 mol% about 5 mol% to about 22 mol% about 10 mol% to about 20 mol%, about 15 mol% to about 20 moltotal iridium relative to the substrate.
  • the catalyst loading is about 20 mol% total iridium.
  • the iridium catalyst comprises a chiral ligand.
  • the asymmetric environment that is created around the metal center by the presence of chiral ligands produces an enantioselective reaction.
  • the chiral ligand forms a complex with the transition metal (i.e., iridium), thereby occupying one or more of the coordination sites on the metal and creating an asymmetric environment around the metal center. This complexation may or may not involve the displacement of achiral ligands already complexed to the metal.
  • the displacement may proceed in a concerted fashion, i.e., with both the achiral ligand decomplexing from the metal and the chiral ligand complexing to the metal in a single step.
  • the displacement may proceed in a stepwise fashion, i.e., with decomplexing of the achiral ligand and complexing of the chiral ligand occurring in distinct steps.
  • Complexation of the chiral ligand to the transition metal may be allowed to occur in situ, i.e., by admixing the ligand and metal before adding the substrate.
  • the ligand-metal complex can be formed separately, and the complex isolated before use in the alkylation reactions of the present invention.
  • the chiral ligand influences the orientation of other molecules as they interact with the transition metal catalyst. Coordination of the metal center with a ⁇ -allyl group and reaction of the substrate with the ⁇ -allyl-metal complex are dictated by the presence of the chiral ligand. The orientation of the reacting species determines the stereochemistry of the products.
  • Chiral ligands of the invention may be bidentate or monodentate or, alternatively, ligands with higher denticity (e.g., tridentate, tetradentate, etc.) can be used.
  • the ligand is a bidentate ligand.
  • substantially enantiopure ligands e.g., ee >99%, preferably ee >99.5%, even more preferably ee >99.9%
  • eee >99%, preferably ee >99.5%, even more preferably ee >99.9% can be purchased from commercial sources, obtained by successive recrystallizations of an enantioenriched substance, or by other suitable means for separating enantiomers.
  • Exemplary chiral ligands may be found in U.S. Patent No. 7,863,443 and CN Patent No. 105524111B .
  • the chiral ligand is an enantioenriched phosphine ligand.
  • the enantioenriched phosphorus-based ligand is a phosphoramidite ligand.
  • the transition metal complex with the ligand comprises S -(CF 3 )- t -BuPHOX, S , S -Et-FerroTANE, S,R p - yliphos, or S,R p -BTFM-xyliphos.
  • the transition metal complex with the ligand comprises R -(CF 3 )- t -BuPHOX, R , R -Et-FerroTANE, R,S p - Xyliphos, or R,S p -BTFM-xyliphos.
  • the chiral ligand is present in an amount in the range of about 1 equivalent to about 20 equivalents relative to the amount of total metal from the catalyst, preferably in the range of about 1 to about 15 equivalents relative to the amount of total metal from the catalyst, and most preferably about 1 equivalent relative to the amount of total metal from the catalyst.
  • the amount of the chiral ligand can be measured relative to the amount of the substrate.
  • the ligand is present under the conditions of the reaction from an amount of about 5 mol% to about 80 mol% relative to the substrate, e.g., the compound of formula (II) or formula (VII).
  • the amount of the chiral ligand present in the reaction is alternatively referred to herein as "ligand loading" and is expressed as a percentage that is calculated by dividing the moles of ligand by the moles of the substrate present in a given reaction.
  • the ligand loading is from about 5 mol%, about 6 mol%, about 7 mol%, about 10 mol%, about 12 mol%, about 14 mol%, about 16 mol%, about 18 mol%, about 19 mol%, about 19.5 mol%, about 19.8 mol%, about 20 mol%, about 20.2 mol%, about 20.5mol%, about 20.8 mol%, about 21 mol%, about 21.2 mol%, about 21.4 mol%, about 21.8 mol%, about 22 mol%, about 25 mol%, about 28 mol%, about 30mol%, about 35 mol%, about 40 mol%, about 45 mol%, about 50 mol%, about 55 mol%, about 58 mol%, about 60 mol%, or about 70 mol%. In preferred embodiments, the ligand loading is 21 mol%.
  • the reactions of the invention may create multiple stereocenters in the product compound, such as the compound of Formula (I), (V) or (VI), in a high degree of enantiomeric excess (ee).
  • the ee of a compound may be measured by dividing the difference in the fractions of the enantiomers by the sum of the fractions of the enantiomers. For example, if a compound is found to comprise 98% (S)-enantiomer, and 2% (R) enantiomer, then the ee of the compound is (98-2)/(98+2), or 96%.
  • the compound of formula (I), (V) or (VI) has about 30% ee or greater, about 40% ee or greater, about 50% ee or greater, 60% ee or greater, about 70% ee or greater, about 80% ee or greater, about 85% ee or greater, about 88% ee or greater, about 90% ee or greater, about 91% ee or greater, about 92% ee or greater, about 93% ee or greater, about 94% ee or greater, about 95% ee or greater, about 96% ee or greater, about 97% ee or greater, about 98% ee or greater, or about 99% ee or greater, even where this % ee is greater than the % ee of the starting material, such as 0% ee (racemic).
  • Methods for treating or preventing cancer comprising administering to a subject in need thereof a therapeutically effective amount of a compound (e.g., a compound of Formula V, VA, VB, VI, VIA, or VIB), or a pharmaceutical composition comprising said compound.
  • a compound e.g., a compound of Formula V, VA, VB, VI, VIA, or VIB
  • a pharmaceutical composition comprising said compound.
  • the cancer that is treated by the methods may be Acute Lymphoblastic Leukemia (ALL), Acute Myeloid Leukemia (AML), Adrenocortical Carcinoma, Anal Cancer, Appendix Cancer, Atypical Teratoid/Rhabdoid Tumor, Basal Cell Carcinoma, Bile Duct Cancer, Bladder Cancer, Bone Cancer, Brain Tumor, Astrocytoma, Brain and Spinal Cord Tumor, Brain Stem Glioma, Central Nervous System Atypical Teratoid/Rhabdoid Tumor, Central Nervous System Embryonal Tumors, Breast Cancer, Bronchial Tumors, Burkitt Lymphoma, Carcinoid Tumor, Carcinoma of Unknown Primary, Central Nervous System Cancer, Cervical Cancer, Childhood Cancers, Chordoma, Chronic Lymphocytic Leukemia (CLL), Chronic Myelogenous Leukemia (CML), Chronic Myeloproliferative Disorders, Colon Cancer, Colorectal Cancer, Cran
  • the cancer that is treated by the methods may be a variety of acute myeloid leukemia (AML), bladder cancer, breast cancer, colorectal cancer, chronic myelogenous leukemia (CML), esophageal cancer, gastric cancer, lung cancer, melanoma, mesothelioma, non-small cell lung carcinoma (NSCLC), ovarian cancer, pancreatic cancer, prostate cancer, renal cancer, or skin cancer.
  • AML acute myeloid leukemia
  • CML chronic myelogenous leukemia
  • NSCLC non-small cell lung carcinoma
  • the cancer that is treated by the may be a variety of acute myeloid leukemia (AML), breast cancer, colorectal cancer, chronic myelogenous leukemia (CML), esophageal cancer, gastric cancer, lung cancer, melanoma, non-small cell lung carcinoma (NSCLC), pancreatic cancer, prostate cancer, or renal cancer.
  • AML acute myeloid leukemia
  • CML chronic myelogenous leukemia
  • NSCLC non-small cell lung carcinoma
  • pancreatic cancer prostate cancer
  • renal cancer renal cancer
  • Combination therapy is an important treatment modality in many disease settings, such as cancer. Recent scientific advances have increased our understanding of the pathophysiological processes that underlie these and other complex diseases. This increased understanding has provided impetus to develop new therapeutic approaches using combinations of drugs directed at multiple therapeutic targets to improve treatment response, minimize development of resistance, or minimize adverse events. In settings in which combination therapy provides significant therapeutic advantages, there is growing interest in the development of combinations with new investigational drugs, such as bis-THIQs.
  • Treating cancer may comprise conjointly administering a chemotherapeutic agent and a compound prepared by the method of the invention.
  • the chemotherapeutic may be an immune-stimulating agent.
  • the immune-stimulating agent may be a pro-inflammatory agent.
  • the chemotherapeutic agent that may be conjointly administered with the bis-THIQs described herein include aminoglutethimide, amsacrine, anastrozole, asparaginase, AZD5363, Bacillus Calmette-Guérin vaccine (beg), bicalutamide, bleomycin, bortezomib, buserelin, busulfan, campothecin, capecitabine, carboplatin, carfilzomib, carmustine, chlorambucil, chloroquine, cisplatin, cladribine, clodronate, cobimetinib, colchicine, cyclophosphamide, cyproterone, cytarabine, dacarbazine, dactinomycin, daunorubicin, demethoxyviridin, dexamethasone, dichloroacetate, dienestrol, diethylstilbestrol, docetaxel, doxorubi
  • the chemotherapeutic agent that may be administered with the bis-THIQs described herein may include abagovomab, adecatumumab, afutuzumab, anatumomab mafenatox, apolizumab, atezolizumab, blinatumomab, catumaxomab, durvalumab, epacadostat, epratuzumab, inotuzumab ozogamicin, intelumumab, ipilimumab, isatuximab, lambrolizumab, nivolumab, ocaratuzumab, olatatumab, pembrolizumab, pidilizumab, ticilimumab, samalizumab, or tremelimumab.
  • the chemotherapeutic agent may be ipilimumab, nivolumab, pembrolizumab, or pidilizumab.
  • combination therapies have been developed for the treatment of cancer.
  • compounds prepared by the method of the invention may be conjointly administered with a combination therapy.
  • Examples of combination therapies with which the compounds may be conjointly administered are included in Table 1.
  • Table 1 Exemplary combinatorial therapies for the treatment of cancer.
  • Immune-targeted agents act against tumors by modulating immune cells.
  • the field of cancer immunotherapy is rapidly growing, with new targets constantly being identified (Chen and Mellman, 2013; Morrissey et al., 2016; Kohrt et al., 2016).
  • immuno-oncology agents comprise agents that modulate immune checkpoints such as 2B4, 4-1BB (CD137), AaR, B7-H3, B7-H4, BAFFR, BTLA, CD2, CD7, CD27, CD28, CD30, CD40, CD80, CD83 ligand, CD86, CD160, CD200, CDS, CEACAM, CTLA-4, GITR, HVEM, ICAM-1, KIR, LAG-3, LAIR1, LFA-1 (CD11a/CD18), LIGHT, NKG2C, NKp80, OX40, PD-1, PD-L1, PD-L2, SLAMF7, TGFR ⁇ , TIGIT, Tim3 and VISTA.
  • Immuno-oncology agents may be in the form of antibodies, peptides, small molecules or viruses.
  • the conjointly administered chemotherapeutic agent may be an immuno-oncology therapeutic agent, such as an inhibitor of arginase, CTLA-4, indoleamine 2,3-dioxygenase, and/or PD-1/PD-L1.
  • the immuno-oncology therapeutic agent may be abagovomab, adecatumumab, afutuzumab, alemtuzumab, anatumomab mafenatox, apolizumab, atezolizumab, avelumab, blinatumomab, BMS-936559, catumaxomab, durvalumab, epacadostat, epratuzumab, indoximod, inotuzumab ozogamicin, intelumumab, ipilimumab, isatuximab, lambrolizumab, MED14736, MPDL3280A, nivoluma
  • the immuno-oncology therapeutic agent is abagovomab, adecatumumab, afutuzumab, anatumomab mafenatox, apolizumab, atezolizumab, blinatumomab, catumaxomab, durvalumab, epacadostat, epratuzumab, indoximod, inotuzumab ozogamicin, intelumumab, ipilimumab, isatuximab, lambrolizumab, nivolumab, ocaratuzumab, olatatumab, pembrolizumab, pidilizumab, ticilimumab, samalizumab, or tremelimumab.
  • immuno-oncology agents are disclosed in Adams, J. L. et al. "Big Opportunities for Small Molecules in Immuno-Oncology” Nature Reviews Drug Discovery 2015, 14, page 603-621 .
  • the conjointly administered chemotherapeutic agent may be a pro-inflammatory agent.
  • the pro-inflammatory agent administered with the bis-THIQs prepared by the method of the invention mya be a cytokine or a chemokine.
  • Chemokines are a group of small cytokines. Pro-inflammatory chemokines promote recruitment and activation of multiple lineages of leukocytes (e.g., lymphocytes, macrophages). Chemokines are related in primary structure and share several conserved amino acid residues. In particular, chemokines typically include two or four cysteine residues that contribute to the three-dimensional structure via formation of disulfide bonds. Chemokines may be classified in one of four groups: C-C chemokines, C-X-C chemokines, C chemokines, and C-X 3 -C chemokines.
  • C-X-C chemokines include a number of potent chemoattractants and activators of neutrophils, such as interleukin 8 (IL-8), PF4 and neutrophil-activating peptide-2 (NAP-2).
  • the C-C chemokines include, for example, RANTES (Regulated on Activation, Normal T Expressed and Secreted), macrophage inflammatory proteins 1-alpha and 1-beta (MIP-1 ⁇ and MIP-1 ⁇ ), eotaxin and human monocyte chemotactic proteins 1 to 3 (MCP-1, MCP-2, MCP-3), which have been characterized as chemoattractants and activators of monocytes or lymphocytes.
  • exemplary pro-inflammatory chemokines include MIP-1 ⁇ , MIP-1 ⁇ , MIP-1 ⁇ , MCP-1, MCP-2, MCP-3, IL-8, PF4, NAP-2, RANTES, CCL2, CCL3, CCL4, CCL5, CCL11, CXCL2, CXCL8, and CXCL10.
  • the method of treating or preventing cancer may further comprise administering one or more non-chemical methods of cancer treatment, such as radiation therapy, surgery, thermoablation, focused ultrasound therapy, cryotherapy, or a combination of the foregoing.
  • one or more non-chemical methods of cancer treatment such as radiation therapy, surgery, thermoablation, focused ultrasound therapy, cryotherapy, or a combination of the foregoing.
  • the chemotherapeutic agent may be administered simultaneously with the bis-THIQ.
  • the chemotherapeutic agent may be administered within about 5 minutes to within about 168 hours prior or after of the bis-THIQ.
  • the step of administering may comprise oral administration of the therapeutic agent.
  • the step of administering can comprise parenteral administration of the therapeutic agent. Further methods of administration are discussed herein.
  • the subject may be a human.
  • the therapeutic agent may be a compound of Formula V, VA, VB, VI, VIA, or VIB. Exemplary compounds are described herein.
  • Also described but not claimed is a method for treating or preventing cancer, comprising conjointly administering to a subject in need thereof a therapeutically effective amount of a compound of Formula V, VA, VB, VI, VIA, or VIB and one or more additional chemotherapeutic agents.
  • acyl is art-recognized and refers to a group represented by the general formula hydrocarbylC(O)-, preferably alkylC(O)-.
  • acylamino is art-recognized and refers to an amino group substituted with an acyl group and may be represented, for example, by the formula hydrocarbylC(O)NH-.
  • acyloxy is art-recognized and refers to a group represented by the general formula hydrocarbylC(O)O-, preferably alkylC(O)O-.
  • alkoxy refers to an alkyl group, preferably a lower alkyl group, having an oxygen attached thereto.
  • Representative alkoxy groups include methoxy, ethoxy, propoxy, tert-butoxy and the like.
  • alkyl group or “alkane” is a straight chained or branched non-aromatic hydrocarbon which is completely saturated. Typically, a straight chained or branched alkyl group has from 1 to about 20 carbon atoms, preferably from 1 to about 10 unless otherwise defined. Examples of straight chained and branched alkyl groups include methyl, ethyl, n-propyl, iso-propyl, n-butyl, sec-butyl, tert-butyl, pentyl, hexyl, pentyl and octyl. A C 1 -C 6 straight chained or branched alkyl group is also referred to as a "lower alkyl" group.
  • alkyl (or “lower alkyl) as used throughout the specification, examples, and claims is intended to include both “unsubstituted alkyls” and “substituted alkyls”, the latter of which refers to alkyl moieties having substituents replacing one or more hydrogens on one or more carbons of the hydrocarbon backbone.
  • substituents can include, for example, a halogen, a hydroxyl, a carbonyl (such as a carboxyl, an alkoxycarbonyl, a formyl, or an acyl), a thiocarbonyl (such as a thioester, a thioacetate, or a thioformate), an alkoxyl, a phosphoryl, a phosphate, a phosphonate, a phosphinate, an amino, an amido, an amidine, an imine, a cyano, a nitro, an azido, a sulfhydryl, an alkylthio, a sulfate, a sulfonate, a sulfamoyl, a sulfonamido, a sulfonyl, a heterocyclyl, an aralkyl, a guanidino, or an aromatic or
  • the moieties substituted on the hydrocarbon chain can themselves be substituted, if appropriate.
  • the substituents of a substituted alkyl may include substituted and unsubstituted forms of amino, azido, imino, amido, phosphoryl (including phosphonate and phosphinate), sulfonyl (including sulfate, sulfonamide, sulfamoyl and sulfonate), and silyl groups, as well as ethers, alkylthios, carbonyls (including ketones, aldehydes, carboxylates, and esters), -CF 3 , -CN and the like.
  • Cycloalkyls can be further substituted with alkyls, alkenyls, alkoxys, alkylthios, aminoalkyls, carbonyl-substituted alkyls, -CF 3 , -CN, and the like.
  • C x-y when used in conjunction with a chemical moiety, such as, acyl, acyloxy, alkyl, alkenyl, alkynyl, or alkoxy is meant to include groups that contain from x to y carbons in the chain.
  • C x-y alkyl refers to substituted or unsubstituted saturated hydrocarbon groups, including straight-chain alkyl and branchedchain alkyl groups that contain from x to y carbons in the chain, including haloalkyl groups such as trifluoromethyl and 2,2,2-trifluoroethyl, etc.
  • C 0 alkyl indicates a hydrogen where the group is in a terminal position, a bond if internal.
  • C 2- y alkenyl and C 2-y alkynyl refer to substituted or unsubstituted unsaturated aliphatic groups analogous in length and possible substitution to the alkyls described above, but that contain at least one double or triple bond respectively.
  • alkylamino refers to an amino group substituted with at least one alkyl group.
  • alkylthio refers to a thiol group substituted with an alkyl group and may be represented by the general formula alkylS-.
  • alkynyl refers to an aliphatic group containing at least one triple bond and is intended to include both "unsubstituted alkynyls" and “substituted alkynyls", the latter of which refers to alkynyl moieties having substituents replacing a hydrogen on one or more carbons of the alkynyl group. Such substituents may occur on one or more carbons that are included or not included in one or more triple bonds. Moreover, such substituents include all those contemplated for alkyl groups, as discussed above, except where stability is prohibitive. For example, substitution of alkynyl groups by one or more alkyl, carbocyclyl, aryl, heterocyclyl, or heteroaryl groups is contemplated.
  • amide refers to a group wherein each R 10 independently represent a hydrogen or hydrocarbyl group, or two R 10 are taken together with the N atom to which they are attached complete a heterocycle having from 4 to 8 atoms in the ring structure.
  • amine and “amino” are art-recognized and refer to both unsubstituted and substituted amines and salts thereof, e.g., a moiety that can be represented by wherein each R 10 independently represents a hydrogen or a hydrocarbyl group, or two R 10 are taken together with the N atom to which they are attached complete a heterocycle having from 4 to 8 atoms in the ring structure.
  • aminoalkyl refers to an alkyl group substituted with an amino group.
  • aralkyl refers to an alkyl group substituted with an aryl group.
  • aryl as used herein include substituted or unsubstituted single-ring aromatic groups in which each atom of the ring is carbon.
  • the ring is a 5- to 7-membered ring, more preferably a 6-membered ring.
  • aryl also includes polycyclic ring systems having two or more cyclic rings in which two or more carbons are common to two adjoining rings wherein at least one of the rings is aromatic, e.g., the other cyclic rings can be cycloalkyls, cycloalkenyls, cycloalkynyls, aryls, heteroaryls, and/or heterocyclyls.
  • Aryl groups include benzene, naphthalene, phenanthrene, phenol, aniline, and the like.
  • carboxylate is art-recognized and refers to a group wherein R 9 and R 10 independently represent hydrogen or a hydrocarbyl group, such as an alkyl group, or R 9 and R 10 taken together with the intervening atom(s) complete a heterocycle having from 4 to 8 atoms in the ring structure.
  • carbocycle refers to a saturated or unsaturated ring in which each atom of the ring is carbon.
  • carbocycle includes both aromatic carbocycles and non-aromatic carbocycles.
  • Non-aromatic carbocycles include both cycloalkane rings, in which all carbon atoms are saturated, and cycloalkene rings, which contain at least one double bond.
  • Carbocycle includes 5-7 membered monocyclic and 8-12 membered bicyclic rings. Each ring of a bicyclic carbocycle may be selected from saturated, unsaturated and aromatic rings.
  • Carbocycle includes bicyclic molecules in which one, two or three or more atoms are shared between the two rings.
  • the term "fused carbocycle” refers to a bicyclic carbocycle in which each of the rings shares two adjacent atoms with the other ring.
  • Each ring of a fused carbocycle may be selected from saturated, unsaturated and aromatic rings.
  • an aromatic ring e.g., phenyl
  • an aromatic ring e.g., phenyl
  • a saturated or unsaturated ring e.g., cyclohexane, cyclopentane, or cyclohexene. Any combination of saturated, unsaturated and aromatic bicyclic rings, as valence permits, is included in the definition of carbocyclic.
  • Carbocycles include cyclopentane, cyclohexane, bicyclo[2.2.1]heptane, 1,5-cyclooctadiene, 1,2,3,4-tetrahydronaphthalene, bicyclo[4.2.0]oct-3-ene, naphthalene and adamantane.
  • Exemplary fused carbocycles include decalin, naphthalene, 1,2,3,4-tetrahydronaphthalene, bicyclo[4.2.0]octane, 4,5,6,7-tetrahydro-1H-indene and bicyclo[4.1.0]hept-3-ene.
  • Carbocycles may be susbstituted at any one or more positions capable of bearing a hydrogen atom.
  • a “cycloalkyl” group is a cyclic hydrocarbon which is completely saturated.
  • “Cycloalkyl” includes monocyclic and bicyclic rings. Typically, a monocyclic cycloalkyl group has from 3 to about 10 carbon atoms, more typically 3 to 8 carbon atoms unless otherwise defined.
  • the second ring of a bicyclic cycloalkyl may be selected from saturated, unsaturated and aromatic rings. Cycloalkyl includes bicyclic molecules in which one, two or three or more atoms are shared between the two rings.
  • the term “fused cycloalkyl” refers to a bicyclic cycloalkyl in which each of the rings shares two adjacent atoms with the other ring.
  • the second ring of a fused bicyclic cycloalkyl may be selected from saturated, unsaturated and aromatic rings.
  • a "cycloalkenyl” group is a cyclic hydrocarbon containing one or more double bonds.
  • (cycloalkyl)alkyl refers to an alkyl group substituted with a cycloalkyl group.
  • carbonate is art-recognized and refers to a group -OCO 2 -R 10 , wherein R 10 represents a hydrocarbyl group.
  • esters refers to a group -C(O)OR 10 wherein R 10 represents a hydrocarbyl group.
  • ether refers to a hydrocarbyl group linked through an oxygen to another hydrocarbyl group. Accordingly, an ether substituent of a hydrocarbyl group may be hydrocarbyl-O-. Ethers may be either symmetrical or unsymmetrical. Examples of ethers include, but are not limited to, heterocycle-O-heterocycle and aryl-O-heterocycle. Ethers include "alkoxyalkyl” groups, which may be represented by the general formula alkyl-O-alkyl.
  • heteroarylkyl refers to an alkyl group substituted with a heteroaryl group.
  • heteroalkyl refers to a saturated or unsaturated chain of carbon atoms and at least one heteroatom, wherein no two heteroatoms are adjacent.
  • heteroaryl includes substituted or unsubstituted aromatic single ring structures, preferably 5- to 7-membered rings, more preferably 5- to 6-membered rings, whose ring structures include at least one heteroatom, preferably one to four heteroatoms, more preferably one or two heteroatoms.
  • heteroaryl also include polycyclic ring systems having two or more cyclic rings in which two or more carbons are common to two adjoining rings wherein at least one of the rings is heteroaromatic, e.g., the other cyclic rings can be cycloalkyls, cycloalkenyls, cycloalkynyls, aryls, heteroaryls, and/or heterocyclyls.
  • Heteroaryl groups include, for example, pyrrole, furan, thiophene, imidazole, oxazole, thiazole, pyrazole, pyridine, pyrazine, pyridazine, and pyrimidine, and the like.
  • heteroatom as used herein means an atom of any element other than carbon or hydrogen. Preferred heteroatoms are nitrogen, oxygen, and sulfur.
  • heterocycloalkyl refers to substituted or unsubstituted non-aromatic ring structures, preferably 3- to 10-membered rings, more preferably 3- to 7-membered rings, whose ring structures include at least one heteroatom, preferably one to four heteroatoms, more preferably one or two heteroatoms.
  • heterocycloalkyl and “heterocyclic” also include polycyclic ring systems having two or more cyclic rings in which two or more carbons are common to two adjoining rings wherein at least one of the rings is heterocyclic, e.g., the other cyclic rings can be cycloalkyls, cycloalkenyls, cycloalkynyls, aryls, heteroaryls, and/or heterocyclyls.
  • Heterocycloalkyl groups include, for example, piperidine, piperazine, pyrrolidine, morpholine, lactones, lactams, and the like.
  • heterocycloalkylalkyl refers to an alkyl group substituted with a heterocycloalkyl group.
  • hydroxyalkyl refers to an alkyl group substituted with a hydroxy group.
  • lower when used in conjunction with a chemical moiety, such as, acyl, acyloxy, alkyl, alkenyl, alkynyl, or alkoxy is meant to include groups where there are ten or fewer non-hydrogen atoms in the substituent, preferably six or fewer.
  • acyl, acyloxy, alkyl, alkenyl, alkynyl, or alkoxy substituents defined herein are respectively lower acyl, lower acyloxy, lower alkyl, lower alkenyl, lower alkynyl, or lower alkoxy, whether they appear alone or in combination with other substituents, such as in the recitations hydroxyalkyl and aralkyl (in which case, for example, the atoms within the aryl group are not counted when counting the carbon atoms in the alkyl substituent).
  • polycyclyl refers to two or more rings (e.g., cycloalkyls, cycloalkenyls, cycloalkynyls, aryls, heteroaryls, and/or heterocyclyls) in which two or more atoms are common to two adjoining rings, e.g., the rings are "fused rings".
  • Each of the rings of the polycycle can be substituted or unsubstituted.
  • each ring of the polycycle contains from 3 to 10 atoms in the ring, preferably from 5 to 7.
  • substituted refers to moieties having substituents replacing a hydrogen on one or more carbons of the backbone. It will be understood that “substitution” or “substituted with” includes the implicit proviso that such substitution is in accordance with permitted valence of the substituted atom and the substituent, and that the substitution results in a stable compound, e.g., which does not spontaneously undergo transformation such as by rearrangement, cyclization, elimination, etc. As used herein, the term “substituted” is contemplated to include all permissible substituents of organic compounds.
  • the permissible substituents include acyclic and cyclic, branched and unbranched, carbocyclic and heterocyclic, aromatic and non-aromatic substituents of organic compounds.
  • the permissible substituents can be one or more and the same or different for appropriate organic compounds.
  • the heteroatoms such as nitrogen may have hydrogen substituents and/or any permissible substituents of organic compounds described herein which satisfy the valences of the heteroatoms.
  • Substituents can include any substituents described herein, for example, a halogen, a hydroxyl, a carbonyl (such as a carboxyl, an alkoxycarbonyl, a formyl, or an acyl), a thiocarbonyl (such as a thioester, a thioacetate, or a thioformate), an alkoxyl, a phosphoryl, a phosphate, a phosphonate, a phosphinate, an amino, an amido, an amidine, an imine, a cyano, a nitro, an azido, a sulfhydryl, an alkylthio, a sulfate, a sulfonate, a sulfamoyl, a sulfonamido, a sulfonyl, a heterocyclyl, an aralkyl, or an aromatic or heteroaromatic mo
  • sulfate is art-recognized and refers to the group -OSO 3 H, or a pharmaceutically acceptable salt thereof.
  • sulfonamide is art-recognized and refers to the group represented by the general formulae wherein R 9 and R 10 independently represents hydrogen or hydrocarbyl, such as alkyl, or R 9 and R 10 taken together with the intervening atom(s) complete a heterocycle having from 4 to 8 atoms in the ring structure.
  • sulfoxide is art-recognized and refers to the group -S(O)-R 10 , wherein R 10 represents a hydrocarbyl.
  • sulfonate is art-recognized and refers to the group SO 3 H, or a pharmaceutically acceptable salt thereof.
  • sulfone is art-recognized and refers to the group -S(O) 2 -R 10 , wherein R 10 represents a hydrocarbyl.
  • thioalkyl refers to an alkyl group substituted with a thiol group.
  • thioester refers to a group -C(O)SR 10 or -SC(O)R 10 wherein R 10 represents a hydrocarbyl.
  • thioether is equivalent to an ether, wherein the oxygen is replaced with a sulfur.
  • urea is art-recognized and may be represented by the general formula wherein R 9 and R 10 independently represent hydrogen or a hydrocarbyl, such as alkyl, or either occurrence of R 9 taken together with R 10 and the intervening atom(s) complete a heterocycle having from 4 to 8 atoms in the ring structure.
  • Protecting group refers to a group of atoms that, when attached to a reactive functional group in a molecule, mask, reduce or prevent the reactivity of the functional group. Typically, a protecting group may be selectively removed as desired during the course of a synthesis. Examples of protecting groups can be found in Greene and Wuts, Protective Groups in Organic Chemistry, 3 rd Ed., 1999, John Wiley & Sons, NY and Harrison et al., Compendium of Synthetic Organic Methods, Vols. 1-8, 1971-1996, John Wiley & Sons, NY.
  • nitrogen protecting groups include, but are not limited to, formyl, acetyl, trifluoroacetyl, benzyl, benzyloxycarbonyl (“CBZ”), tertbutoxycarbonyl (“Boc”), trimethylsilyl (“TMS”), 2-trimethylsilyl-ethanesulfonyl (“TES”), trityl and substituted trityl groups, allyloxycarbonyl, 9-fluorenylmethyloxycarbonyl (“FMOC”), nitro-veratryloxycarbonyl (“NVOC”) and the like.
  • hydroxyl protecting groups include, but are not limited to, those where the hydroxyl group is either acylated (esterified) or alkylated such as benzyl and trityl ethers, as well as alkyl ethers, tetrahydropyranyl ethers, trialkylsilyl ethers (e.g., TMS or TIPS groups), glycol ethers, such as ethylene glycol and propylene glycol derivatives and allyl ethers.
  • a therapeutic that "prevents" a disorder or condition refers to a compound that, in a statistical sample, reduces the occurrence of the disorder or condition in the treated sample relative to an untreated control sample, or delays the onset or reduces the severity of one or more symptoms of the disorder or condition relative to the untreated control sample.
  • treating includes prophylactic and/or therapeutic treatments.
  • prophylactic or therapeutic treatment is art-recognized and includes administration to the host of one or more of the subject compositions. If it is administered prior to clinical manifestation of the unwanted condition (e.g., disease or other unwanted state of the host animal) then the treatment is prophylactic (i.e., it protects the host against developing the unwanted condition), whereas if it is administered after manifestation of the unwanted condition, the treatment is therapeutic, (i.e., it is intended to diminish, ameliorate, or stabilize the existing unwanted condition or side effects thereof).
  • prodrug is intended to encompass compounds which, under physiologic conditions, are converted into the therapeutically active agents (e.g., a compound of Formula V, VA, VB, VI, VIA, or VIB).
  • a common method for making a prodrug is to include one or more selected moieties which are hydrolyzed under physiologic conditions to reveal the desired molecule.
  • the prodrug may be converted by an enzymatic activity of the host animal.
  • esters or carbonates e.g., esters or carbonates of alcohols or carboxylic acids
  • amides e.g., an amide of an amino group
  • Some or all of the active compounds in a formulation represented above can be replaced with the corresponding suitable prodrug, e.g., wherein a hydroxyl in the parent compound is presented as an ester or a carbonate or carboxylic acid present in the parent compound is presented as an ester.
  • a pharmaceutical composition is described but not claimed comprising a compound described above (e.g., a compound of Formula V, VA, VB, VI, VIA, or VIB), or a pharmaceutically acceptable salt thereof; and a pharmaceutically acceptable carrier.
  • a pharmaceutical preparation suitable for use in a human patient comprising any compound described above (e.g., a compound of Formula V, VA, VB, VI, VIA, or VIB), and one or more pharmaceutically acceptable excipients.
  • the pharmaceutical preparations may be for use in treating or preventing a condition or disease as described herein.
  • the pharmaceutical preparations may have a low enough pyrogen activity to be suitable for use in a human patient.
  • a pharmaceutical kit comprising a compound described above (e.g., a compound of Formula V, VA, VB, VI, VIA, or VIB), or a pharmaceutically acceptable salt thereof, and optionally directions on how to administer the compound.
  • compositions and methods may be utilized to treat an individual in need thereof.
  • the individual may be a mammal such as a human, or a non-human mammal.
  • the composition or the compound When administered to an animal, such as a human, the composition or the compound is preferably administered as a pharmaceutical composition comprising, for example, the compound and a pharmaceutically acceptable carrier.
  • Pharmaceutically acceptable carriers are well known in the art and include, for example, aqueous solutions such as water or physiologically buffered saline or other solvents or vehicles such as glycols, glycerol, oils such as olive oil, or injectable organic esters.
  • the aqueous solution may be pyrogen-free, or substantially pyrogen-free.
  • the excipients can be chosen, for example, to effect delayed release of an agent or to selectively target one or more cells, tissues or organs.
  • the pharmaceutical composition can be in dosage unit form such as tablet, capsule (including sprinkle capsule and gelatin capsule), granule, lyophile for reconstitution, powder, solution, syrup, suppository, injection or the like.
  • the composition can also be present in a transdermal delivery system, e.g., a skin patch.
  • the composition can also be present in a solution suitable for topical administration, such as an eye drop.
  • a pharmaceutically acceptable carrier can contain physiologically acceptable agents that act, for example, to stabilize, increase solubility or to increase the absorption of a compound such as a compound described above.
  • physiologically acceptable agents include, for example, carbohydrates, such as glucose, sucrose or dextrans, antioxidants, such as ascorbic acid or glutathione, chelating agents, low molecular weight proteins or other stabilizers or excipients.
  • the choice of a pharmaceutically acceptable carrier, including a physiologically acceptable agent depends, for example, on the route of administration of the composition.
  • the preparation or pharmaceutical composition can be a selfemulsifying drug delivery system or a selfmicroemulsifying drug delivery system.
  • the pharmaceutical composition also can be a liposome or other polymer matrix, which can have incorporated therein, for example, a compound described above.
  • Liposomes for example, which comprise phospholipids or other lipids, are nontoxic, physiologically acceptable and metabolizable carriers that are relatively simple to make and administer.
  • phrases "pharmaceutically acceptable” is employed herein to refer to those compounds, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio.
  • pharmaceutically acceptable carrier means a pharmaceutically acceptable material, composition or vehicle, such as a liquid or solid filler, diluent, excipient, solvent or encapsulating material. Each carrier must be “acceptable” in the sense of being compatible with the other ingredients of the formulation and not injurious to the patient.
  • materials which can serve as pharmaceutically acceptable carriers include: (1) sugars, such as lactose, glucose and sucrose; (2) starches, such as corn starch and potato starch; (3) cellulose, and its derivatives, such as sodium carboxymethyl cellulose, ethyl cellulose and cellulose acetate; (4) powdered tragacanth; (5) malt; (6) gelatin; (7) talc; (8) excipients, such as cocoa butter and suppository waxes; (9) oils, such as peanut oil, cottonseed oil, safflower oil, sesame oil, olive oil, corn oil and soybean oil; (10) glycols, such as propylene glycol; (11) polyols, such as glycerin, sorbitol, mannitol and polyethylene glycol; (12) esters, such as ethyl oleate and ethyl laurate; (13) agar; (14) buffering agents, such as magnesium hydroxide and aluminum hydroxide;
  • a pharmaceutical composition can be administered to a subject by any of a number of routes of administration including, for example, orally (for example, drenches as in aqueous or non-aqueous solutions or suspensions, tablets, capsules (including sprinkle capsules and gelatin capsules), boluses, powders, granules, pastes for application to the tongue); absorption through the oral mucosa (e.g., sublingually); anally, rectally or vaginally (for example, as a pessary, cream or foam); parenterally (including intramuscularly, intravenously, subcutaneously or intrathecally as, for example, a sterile solution or suspension); nasally; intraperitoneally; subcutaneously; transdermally (for example as a patch applied to the skin); and topically (for example, as a cream, ointment or spray applied to the skin, or as an eye drop).
  • routes of administration including, for example, orally (for example, drenches as in aqueous or
  • the compound may also be formulated for inhalation.
  • the compound may be simply dissolved or suspended in sterile water. Details of appropriate routes of administration and compositions suitable for same can be found in, for example, U.S. Pat. Nos. 6,110,973 , 5,763,493 , 5,731,000 , 5,541,231 , 5,427,798 , 5,358,970 and 4,172,896 , as well as in patents cited therein.
  • the formulations may conveniently be presented in unit dosage form and may be prepared by any methods well known in the art of pharmacy.
  • the amount of active ingredient which can be combined with a carrier material to produce a single dosage form will vary depending upon the host being treated, the particular mode of administration.
  • the amount of active ingredient that can be combined with a carrier material to produce a single dosage form will generally be that amount of the compound which produces a therapeutic effect. Generally, out of one hundred percent, this amount will range from about 1 percent to about ninety-nine percent of active ingredient, preferably from about 5 percent to about 70 percent, most preferably from about 10 percent to about 30 percent.
  • Methods of preparing these formulations or compositions include the step of bringing into association an active compound, such as a compound described above, with the carrier and, optionally, one or more accessory ingredients.
  • an active compound such as a compound described above
  • the formulations are prepared by uniformly and intimately bringing into association the compound with liquid carriers, or finely divided solid carriers, or both, and then, if necessary, shaping the product.
  • Formulations suitable for oral administration may be in the form of capsules (including sprinkle capsules and gelatin capsules), cachets, pills, tablets, lozenges (using a flavored basis, usually sucrose and acacia or tragacanth), lyophile, powders, granules, or as a solution or a suspension in an aqueous or non-aqueous liquid, or as an oil-in-water or water-in-oil liquid emulsion, or as an elixir or syrup, or as pastilles (using an inert base, such as gelatin and glycerin, or sucrose and acacia) and/or as mouth washes and the like, each containing a predetermined amount of the compound as an active ingredient.
  • Compositions or compounds may also be administered as a bolus, electuary or paste.
  • the active ingredient is mixed with one or more pharmaceutically acceptable carriers, such as sodium citrate or dicalcium phosphate, and/or any of the following: (1) fillers or extenders, such as starches, lactose, sucrose, glucose, mannitol, and/or silicic acid; (2) binders, such as, for example, carboxymethylcellulose, alginates, gelatin, polyvinyl pyrrolidone, sucrose and/or acacia; (3) humectants, such as glycerol; (4) disintegrating agents, such as agar-agar, calcium carbonate, potato or tapioca starch, alginic acid, certain silicates, and sodium carbonate; (5) solution retarding agents, such as paraffin; (6) absorption accelerators, such as quaternary ammonium compounds; (7) wetting agents,
  • pharmaceutically acceptable carriers such as sodium citrate or dicalcium phosphate, and/or any of the following: (1) fillers or extenders, such as starches, lactose
  • compositions may also comprise buffering agents.
  • Solid compositions of a similar type may also be employed as fillers in soft and hard-filled gelatin capsules using such excipients as lactose or milk sugars, as well as high molecular weight polyethylene glycols and the like.
  • a tablet may be made by compression or molding, optionally with one or more accessory ingredients.
  • Compressed tablets may be prepared using binder (for example, gelatin or hydroxypropylmethyl cellulose), lubricant, inert diluent, preservative, disintegrant (for example, sodium starch glycolate or cross-linked sodium carboxymethyl cellulose), surface-active or dispersing agent.
  • Molded tablets may be made by molding in a suitable machine a mixture of the powdered compound moistened with an inert liquid diluent.
  • the tablets, and other solid dosage forms of the pharmaceutical compositions may optionally be scored or prepared with coatings and shells, such as enteric coatings and other coatings well known in the pharmaceutical-formulating art. They may also be formulated so as to provide slow or controlled release of the active ingredient therein using, for example, hydroxypropylmethyl cellulose in varying proportions to provide the desired release profile, other polymer matrices, liposomes and/or microspheres.
  • compositions may be sterilized by, for example, filtration through a bacteriaretaining filter, or by incorporating sterilizing agents in the form of sterile solid compositions that can be dissolved in sterile water, or some other sterile injectable medium immediately before use.
  • These compositions may also optionally contain opacifying agents and may be of a composition that they release the active ingredient(s) only, or preferentially, in a certain portion of the gastrointestinal tract, optionally, in a delayed manner.
  • embedding compositions that can be used include polymeric substances and waxes.
  • the active ingredient can also be in micro-encapsulated form, if appropriate, with one or more of the above-described excipients.
  • Liquid dosage forms useful for oral administration include pharmaceutically acceptable emulsions, lyophiles for reconstitution, microemulsions, solutions, suspensions, syrups and elixirs.
  • the liquid dosage forms may contain inert diluents commonly used in the art, such as, for example, water or other solvents, cyclodextrins and derivatives thereof, solubilizing agents and emulsifiers, such as ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propylene glycol, 1,3-butylene glycol, oils (in particular, cottonseed, groundnut, corn, germ (e.g., wheat germ), olive, castor and sesame oils), glycerol, tetrahydrofuryl alcohol, polyethylene glycols and fatty acid esters of sorbitan, and mixtures thereof.
  • Formulations of the pharmaceutical compositions for administration to the mouth may be presented as a mouthwash, or an oral spray, or an oral ointment.
  • compositions can be formulated for delivery via a catheter, stent, wire, or other intraluminal device. Delivery via such devices may be especially useful for delivery to the bladder, urethra, ureter, rectum, or intestine.
  • Formulations which are suitable for vaginal administration also include pessaries, tampons, creams, gels, pastes, foams or spray formulations containing such carriers as are known in the art to be appropriate.
  • Dosage forms for the topical or transdermal administration include powders, sprays, ointments, pastes, creams, lotions, gels, solutions, patches and inhalants.
  • the active compound may be mixed under sterile conditions with a pharmaceutically acceptable carrier, and with any preservatives, buffers, or propellants that may be required.
  • the ointments, pastes, creams and gels may contain, in addition to an active compound, excipients, such as animal and vegetable fats, oils, waxes, paraffins, starch, tragacanth, cellulose derivatives, polyethylene glycols, silicones, bentonites, silicic acid, talc and zinc oxide, or mixtures thereof.
  • excipients such as animal and vegetable fats, oils, waxes, paraffins, starch, tragacanth, cellulose derivatives, polyethylene glycols, silicones, bentonites, silicic acid, talc and zinc oxide, or mixtures thereof.
  • Powders and sprays can contain, in addition to an active compound, excipients such as lactose, talc, silicic acid, aluminum hydroxide, calcium silicates and polyamide powder, or mixtures of these substances.
  • Sprays can additionally contain customary propellants, such as chlorofluorohydrocarbons and volatile unsubstituted hydrocarbons, such as butane and propane.
  • Transdermal patches have the added advantage of providing controlled delivery of a compound to the body.
  • dosage forms can be made by dissolving or dispersing the active compound in the proper medium.
  • Absorption enhancers can also be used to increase the flux of the compound across the skin. The rate of such flux can be controlled by either providing a rate controlling membrane or dispersing the compound in a polymer matrix or gel.
  • Ophthalmic formulations eye ointments, powders, solutions and the like, are also contemplated.
  • Exemplary ophthalmic formulations are described in U.S. Publication Nos. 2005/0080056 , 2005/0059744 , 2005/0031697 and 2005/004074 and U.S. Patent No. 6,583,124 .
  • liquid ophthalmic formulations have properties similar to that of lacrimal fluids, aqueous humor or vitreous humor or are compatable with such fluids.
  • a preferred route of administration is local administration (e.g., topical administration, such as eye drops, or administration via an implant).
  • parenteral administration and “administered parenterally” as used herein means modes of administration other than enteral and topical administration, usually by injection, and includes, without limitation, intravenous, intramuscular, intraarterial, intrathecal, intracapsular, intraorbital, intracardiac, intradermal, intraperitoneal, transtracheal, subcutaneous, subcuticular, intraarticular, subcapsular, subarachnoid, intraspinal and intrasternal injection and infusion.
  • compositions suitable for parenteral administration comprise one or more active compounds in combination with one or more pharmaceutically acceptable sterile isotonic aqueous or nonaqueous solutions, dispersions, suspensions or emulsions, or sterile powders which may be reconstituted into sterile injectable solutions or dispersions just prior to use, which may contain antioxidants, buffers, bacteriostats, solutes which render the formulation isotonic with the blood of the intended recipient or suspending or thickening agents.
  • compositions may also contain adjuvants such as preservatives, wetting agents, emulsifying agents and dispersing agents. Prevention of the action of microorganisms may be ensured by the inclusion of various antibacterial and antifungal agents, for example, paraben, chlorobutanol, phenol sorbic acid, and the like. It may also be desirable to include isotonic agents, such as sugars, sodium chloride, and the like into the compositions. In addition, prolonged absorption of the injectable pharmaceutical form may be brought about by the inclusion of agents that delay absorption such as aluminum monostearate and gelatin.
  • Methods of introduction may also be provided by rechargeable or biodegradable devices.
  • Various slow release polymeric devices have been developed and tested in vivo in recent years for the controlled delivery of drugs, including proteinacious biopharmaceuticals.
  • a variety of biocompatible polymers including hydrogels, including both biodegradable and non-degradable polymers, can be used to form an implant for the sustained release of a compound at a particular target site.
  • the selected dosage level will depend upon a variety of factors including the activity of the particular compound or combination of compounds employed, or the ester, salt or amide thereof, the route of administration, the time of administration, the rate of excretion of the particular compound(s) being employed, the duration of the treatment, other drugs, compounds and/or materials used in combination with the particular compound(s) employed, the age, sex, weight, condition, general health and prior medical history of the patient being treated, and like factors well known in the medical arts.
  • a physician or veterinarian having ordinary skill in the art can readily determine and prescribe the therapeutically effective amount of the pharmaceutical composition required.
  • the physician or veterinarian could start doses of the pharmaceutical composition or compound at levels lower than that required in order to achieve the desired therapeutic effect and gradually increase the dosage until the desired effect is achieved.
  • therapeutically effective amount is meant the concentration of a compound that is sufficient to elicit the desired therapeutic effect. It is generally understood that the effective amount of the compound will vary according to the weight, sex, age, and medical history of the subject. Other factors which influence the effective amount may include, but are not limited to, the severity of the patient's condition, the disorder being treated, the stability of the compound, and, if desired, another type of therapeutic agent being administered with the compound.
  • a larger total dose can be delivered by multiple administrations of the agent.
  • Methods to determine efficacy and dosage are known to those skilled in the art ( Isselbacher et al. (1996) Harrison's Principles of Internal Medicine 13 ed., 1814-1882 ).
  • a suitable daily dose of an active compound used in the compositions and methods described herein will be that amount of the compound that is the lowest dose effective to produce a therapeutic effect. Such an effective dose will generally depend upon the factors described above.
  • the effective daily dose of the active compound may be administered as one, two, three, four, five, six or more sub-doses administered separately at appropriate intervals throughout the day, optionally, in unit dosage forms.
  • the active compound may be administered two or three times daily.
  • the active compound may be administered once daily.
  • the patient receiving this treatment is any animal in need, including primates, in particular humans, and other mammals such as equines, cattle, swine and sheep; and poultry and pets in general.
  • Conjoint administration of compounds prepared by the method of the invention with one or more additional therapeutic agent(s) may provide improved efficacy relative to each individual administration of the compound (e.g., a compound of Formula V, VA, VB, VI, VIA, or VIB) or the one or more additional therapeutic agent(s).
  • the conjoint administration may provide an additive effect, wherein an additive effect refers to the sum of each of the effects of individual administration of the compound and the one or more additional therapeutic agent(s).
  • pharmaceutically acceptable salt includes salts derived from inorganic or organic acids including, for example, hydrochloric, hydrobromic, sulfuric, nitric, perchloric, phosphoric, formic, acetic, lactic, maleic, fumaric, succinic, tartaric, glycolic, salicylic, citric, methanesulfonic, benzenesulfonic, benzoic, malonic, trifluoroacetic, trichloroacetic, naphthalene-2-sulfonic, oxalic, mandelic and other acids.
  • compositions can include forms wherein the ratio of molecules comprising the salt is not 1:1.
  • the salt may comprise more than one inorganic or organic acid molecule per molecule of base, such as two hydrochloric acid molecules per molecule of compound of formula V, VA, VB, VI, VIA, or VIB.
  • the salt may comprise less than one inorganic or organic acid molecule per molecule of base, such as two molecules of compound of Formula V, VA, VB, VI, VIA, or VIB per molecule of tartaric acid.
  • Contemplated salts include, but are not limited to, alkyl, dialkyl, trialkyl or tetraalkyl ammonium salts.
  • contemplated salts of the invention include, but are not limited to, L-arginine, benenthamine, benzathine, betaine, calcium hydroxide, choline, deanol, diethanolamine, diethylamine, 2-(diethylamino)ethanol, ethanolamine, ethylenediamine, N-methylglucamine, hydrabamine, 1H-imidazole, lithium, L-lysine, magnesium, 4-(2-hydroxyethyl)morpholine, piperazine, potassium, 1-(2-hydroxyethyl)pyrrolidine, sodium, triethanolamine, tromethamine, and zinc salts.
  • Contemplated salts may include, but are not limited to, Na, Ca, K, Mg, Zn or other metal salts.
  • the pharmaceutically acceptable acid addition salts can also exist as various solvates, such as with water, methanol, ethanol, dimethylformamide, and the like. Mixtures of such solvates can also be prepared.
  • the source of such solvate can be from the solvent of crystallization, inherent in the solvent of preparation or crystallization, or adventitious to such solvent.
  • wetting agents such as sodium lauryl sulfate and magnesium stearate, as well as coloring agents, release agents, coating agents, sweetening, flavoring and perfuming agents, preservatives and antioxidants can also be present in the compositions.
  • Boekelheide rearrangement 33,34 would be particularly well suited for the advance-ment of our synthesis, utilizing the oxidation already present in the molecule.
  • This species can then undergo a double Boekelheide rearrangement in refluxing acetic anhydride, transmuting the N-oxides to the benzylic acetates.
  • Table 1 above reports the results of the development of the enantioselective hydrogenation. Unless otherwise noted, all reac-tions were run in 9:1 toluene:acetic acid (0.02 M) in the presence of tetra-n-butylammonium iodide (an iodide-to-iridium ratio of 3:1 was maintained in all cases) under a pressurized (60 bar) hydro-gen atmosphere for 18 hr. a Measured by absorption at 230 nm on UHPLC-MS vs. 1,2,4,5-tetrachlorobenzene internal standard unless otherwise noted. b Measured by 1 H-NMR analysis of the crude reaction mixture.
  • ee is enantiomeric excess
  • cod is 1,5-cyclooctadiene
  • TBAI is tetra-n-butylammonium iodide
  • ND is not determined
  • BTFM is bis-trifluoromethyl.
  • IR Infrared
  • HRMS High resolution mass spectra
  • the flask was re-submerged in a -20 °C bath, and freshly distilled methyl iodide (2.25 mL, 36.1 mmol, 6 equiv) was added in a dropwise fashion, resulting in a mild exotherm.
  • the solution was stirred 30 min at -20 °C and was removed from its bath, warming to room temperature. After 30 min the reaction was quenched by the addition of 20 mL 0.5 M HCl, and the solution was stirred 30 min without a cap.
  • the layers were separated and the aqueous phase was saturated with sodium chloride.
  • the aqueous phase was extracted with Et 2 O, dried over MgSO 4 and concentrated.
  • the product was purified by column chromatography (10% EtOAc/hex).
  • O-tert- butyldimethylsilyl propargyl alcohol iii (12, 17.3 g, 101 mmol, 1.35 equiv) was added in one portion, causing the suspension to darken as the palladium catalyst was reduced.
  • the suspension was sparged with N 2 for a further 1 min, then heated to 70 °C for 24 h.
  • TLC and LCMS indicated complete conversion of bromide 11, so the suspension was cooled to 50 °C and 200 mL MeOH was added.
  • Hydroxylamine hydrochloride (6.24 g, 89.8 mmol, 1.2 equiv) was added in one portion and the solution was heated to reflux (85 °C) for 2 h.
  • Triethylamine (4.7 mL, 33.7 mmol, 5 equiv) and isopropyl isocyanate (2.6 mL, 26.9 mmol, 4 equiv) were added at 23 °C and the solution was stirred 16 h, at which time TLC (50% EtOAc/hex) indicated complete conversion to carbamate S4.
  • TLC 50% EtOAc/hex
  • the contents of the vial were transferred to a 100 mL roundbottom flask and 10% aq. Na 2 S 2 O 3 was added to quench the remaining oxidant and citric acid hydrate (4.5 g, >3 equiv) was added to chelate the boron.
  • Trimethylsilyl chloride (61 mL, 478 mmol, 7 equiv) was then added dropwise via the addition funnel over the course of 30 min and the suspension was stirred at -78 °C for 30 min, then was removed from the dry ice bath and stirred at 23 °C for 16 h.
  • the reaction was quenched by the addition of 300 mL aqueous NH 4 Cl (30 mL saturated solution diluted to 300 mL) through an addition funnel, the first 50 mL of which were added dropwise, followed by the addition of the remainder in a slow stream.
  • the aqueous phase was then further acidified by the addition of small portions of concentrated HCl until an acidic pH was achieved ( ⁇ 30 mL required).
  • N -Phenyl triflimide (10.6 g, 29.8 mmol, 1.2 equiv) was added in one portion and the solution was stirred 30 min.
  • a second portion of diethylamine (4.6 mL, 44.7 mmol, 1.8 equiv) was added and the solution was stirred 2 h.
  • the solution was filtered through a 1 inch pad of silica gel with 50% Et 2 O/hex and concentrated.
  • the product was purified by column chromatography (10% Et 2 O/hex).
  • Arene 14 can be isolated as a colorless oil, but undergoes decomposition and should be used within the day of its isolation. Colorless oil, 9.15 g, 24.6 mmol, 99% yield.
  • Cesium fluoride (204 mg, 1.34 mmol, 2.5 equiv) was dissolved in acetonitrile (5.4 mL, 0.1 M) in a 20 mL microwave vial and water (9.7 ⁇ L, 0.537 mmol, 1.0 equiv) and methyl acetoacetate (58 ⁇ L, 0.537 mmol, 1.0 equiv) were added.
  • Aryne precursor 14 250 mg, 0.671 mmol, 1.25 equiv was added neat via syringe, and the vial was placed in a preheated 80 °C oil bath.
  • the aqueous phase was then brought back to pH 7 by the addition of 100 mL 2M NaOH and was extracted with EtOAc (5x 20 mL). The combined organic phases were washed with brine, dried over Na 2 SO 4 and concentrated, providing the product. Yellow solid, 56.9 mg, 0.243 mmol, 45% yield.
  • Isoquinoline triflate 10 (2.77 g, 6.82 mmol, 1.00 equiv) was dissolved in 10 mL toluene, which was sparge-degassed with nitrogen for 10 min.
  • the solution of isoquinoline triflate 10 was then added via cannula to the cooled catalyst solution, rinsing the flask with 5 mL degassed toluene.
  • the catalyst/triflate solution was then added via cannula to the 250 mL sealable flask, rinsing with 10 mL degassed toluene.
  • the flask was sealed and placed in a 130 °C preheated oil bath for 4.5 h.
  • Bis-isoquinoline S8 (5.44 g, 9.7 mmol, 1.00 equiv) was dissolved in acetic acid (40 mL, 0.25 M) and solid potassium fluoride (2.81 g, 48.0 mmol, 5.00 equiv) was added in one portion. The solution was stirred 30 min at room temperature, at which time LCMS showed complete conversion to the product. The solution was diluted with CH 2 Cl 2 and ice and the solution was stirred vigorously as a solution of sodium hydroxide (25 g, 0.625 mol, 0.9 equiv relative to 40 mL AcOH) in 70 mL water was added slowly.
  • bis-N-oxide 19 was not stable to Na 2 SO 4 , MgSO 4 , or SiO 2 , and as such it was neither dried nor purified by column chromatography. The solution was concentrated, providing the analytically pure bis-N-oxide. Red solid, 100.7 mg, 0.169 mmol, 98% yield.
  • Tetra-n-butylammonium iodide (238 mg, 0.644 mmol, 0.6 equiv, 3 equiv relative to Ir) was added to the flask. and this solution was added to the bis-isoquinoline slurry, resulting in a yellow solution of protonated 22. was suspended in 20 mL PhMe (22 is not fully soluble in PhMe alone).
  • [Ir(cod)Cl] 2 (72.1 mg, 0.107 mmol, 0.1 equiv, 20 mol% Ir) and BTFM-Xyliphos (a.k.a.
  • the bomb was removed from the oil bath and the hydrogen pressure was vented.
  • the flask was removed from the bomb and the solution was transferred to a 250 mL roundbottom flask and basified by the careful addition of saturated aqueous K 2 CO 3 and water until pH > 7.
  • the solution was transferred to a separatory funnel and the layers were separated.
  • the aqueous phase was extracted 5x with EtOAc, and the combined organic phases were washed twice with water and once with brine, dried over Na 2 SO 4 , and concentrated.
  • the product was purified by column chromatography (15x1", 1% MeOH/DCM + 1% NEts). At this stage, 1 H NMR determined the purity of the product to be 90% as a brown foam.
  • the resulting crystals were dried in vacuo, providing 203 mg of enantiopure (>99% ee) bis-tetrahydroisoquinoline 6.
  • the crystals isolated above were used to collect the following characterization data.
  • Enantiopure bis-tetrahydroisoquinoline 6 (125 mg, 0.267 mmol, 1 equiv) was dissolved in 1,2-dichloroethane (1,2-DCE, 5.3 mL, 0.05 M) and 37% aqueous formaldehyde (100 ⁇ L, 1.33 mmol, 5 equiv) was added. The solution was stirred at 800 rpm for 10 min, before sodium triacetoxyborohydride (565 mg, 2.67 mmol, 10 equiv) was added. This solution was stirred at 23 °C for 30 min, at which time LCMS showed full conversion to the product.
  • Citric acid monohydrate (840 mg, 4.00 mmol, 15 equiv) was added to the solution, followed by 20 mL water. This solution was stirred for 10 min before the slow addition of saturated aqueous K 2 CO 3 until pH > 7. The layers were separated and the aqueous phase was extracted with CH 2 Cl 2 . The combined organic phases were washed with brine, dried over Na 2 SO 4 and concentrated. The product was purified by column chromatography (1% MeOH/DCM + 1% NEt 3 ). Colorless solid, 118.5 mg, 0.246 mmol, 92% yield.
  • N-Chlorosaccharine (87.7 mg, 0.403 mmol, 2.20 equiv) was dissolved in 1 mL HFIP and this solution was added at a slow dropwise pace, allowing the orange color to dispel after each addition, and the resulting yellow solution was stirred at 0 °C.
  • An LCMS sample taken 5 min after complete addition showed complete dichlorination, so the reaction was quenched by the addition of saturated aqueous Na 2 S 2 O 3 .
  • the resulting mixture was transferred to a separatory funnel with and diluted with CH 2 Cl 2 , creating a triphasic system with HFIP on bottom, CH 2 Cl 2 on the bottom, and the aqueous phase on top.
  • the bottom two phases were collected directly in a 250 mL roundbottom flask.
  • the aqueous phase was basified with K 2 CO 3 and extracted with CH 2 Cl 2 , draining the organic phase directly into the flask.
  • Excess acetic acid ( ⁇ 100 ⁇ L) was added and the solution was concentrated, removing excess acetic acid by azeotropic drying with toluene, and the resulting foam was dried at ⁇ 1 torr for 1 h.
  • the crude product was dissolved in CH 2 Cl 2 and washed with dilute aqueous K 2 CO 3 and the layers were separated.
  • the aqueous phase was extracted with CH 2 Cl 2 and the combined organic phases were dried over Na 2 SO 4 and concentrated.
  • the vial was sealed with electrical tape and removed from the glovebox, sonicated briefly, and returned to the glovebox.
  • the resulting tan solution was then transferred to a 20 mL microwave vial containing bis-tetrahydroisoquinoline 28 (50.0 mg, 0.0907 mmol, 1.00 equiv) and CsOH•H 2 O (152.3 mg, 0.907 mmol, 10.0 equiv), followed by a 1 mL rinse (9.1 mL total volume, 0.01 M in 28 ).
  • the vial was sealed, removed from the glovebox, and placed in a preheated 90 °C oil bath. After 3 h, the vial was removed and allowed to cool fully to room temperature prior to removing the seal.
  • reaction vessel If the reaction vessel is prematurely exposed to air at elevated tempearture, aerobic oxidation leads to the formation of quinones, which undergo hydrolysis of the vinylogous ester in the presence of CsOH.
  • the solution must be fully cooled to room temperature prior to breaking the seal.
  • the bisphenol product is not sensitive to aerobic oxidation, in the solid state or in solution.
  • Acetic acid (46.5 ⁇ L, 0.813 mmol, 9 equiv) was added to quench remaining CsOH and the contents of the vial were transferred to a roundbottom flask, to which silica gel was added directly to dry load onto a silica gel column.
  • the flask was sealed with a rubber septum that was then pierced with three 16 gauge (purple) needles, each bent at a 90° angle.
  • the flask was placed inside the bomb, which was then sealed prior to removal from the glovebox via the large antechamber.
  • the tape was removed from the top of the bomb and the pressure gauge was quickly screwed in place and tightened.
  • the bomb was charged to 10 bar of H 2 and slowly released. This process was repeated twice, before charging the bomb to 60 bar of H 2 , at which time it was placed in a preheated 60°C oil bath. The bath was maintained at this temperature for 18 h, then raised to 80 °C for 24 h.
  • the bomb was removed from the oil bath and the hydrogen pressure was vented.
  • the flask was removed from the bomb and the solution was transferred to a 250 mL roundbottom flask and basified by the careful addition of saturated aqueous K 2 CO 3 and water until pH > 7.
  • the solution was transferred to a separatory funnel and the layers were separated.
  • the aqueous phase was extracted 5x with EtOAc, and the combined organic phases were washed twice with water and once with brine, dried over Na 2 SO 4 , and concentrated.
  • the crude tetrahydroisoquinoline was dissolved in 3 mL THF and triethylamine (3 equiv), DMAP (0.1 equiv), and benzoyl chloride (3 equiv) were added.
  • ( S )-1-(( R P )-2-bis(3,5-bis(trifluoromethyl)phenyl)phosphinoferrocen-1-yl)-1-dimethylamino-ethane (S7) In a flame-dried 2-neck roundbottom flask equipped with a reflux condenser, ( S )-1-ferrocenyldimethylaminoethane (S6) (2.18 g, 8.46 mmol, 1 equiv) was dissolved in diethyl ether (85 mL, 0.1 M) and the solution was cooled to -78 °C.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Organic Low-Molecular-Weight Compounds And Preparation Thereof (AREA)
  • Nitrogen Condensed Heterocyclic Rings (AREA)

Claims (9)

  1. Ein Verfahren zur Herstellung einer Verbindung der Formel (I):
    Figure imgb0081
    beinhaltend das In-Kontakt-Bringen einer Verbindung der Formel (II):
    Figure imgb0082
    mit einem Übergangsmetallkatalysator (vorzugsweise einem chiralen Übergangsmetallkatalysator) unter Hydrierungsbedingungen, wobei, solange Valenz und Stabilität es zulassen:
    R1 und R7 jeweils unabhängig Wasserstoff, Hydroxyl, Halogen, Nitro, Alkyl, Alkenyl, Alkinyl, Cyan, Carboxyl, Sulfat, Amino, Alkoxy, Alkylamino, Alkylthio, Ether, Thioether, Ester, Amid, Thioester, Carbonat, Carbamat, Harnstoff, Sulfonat, Sulfon, Sulfoxid, Sulfonamid, Acyl, Acyloxy, Trialkylsilyloxy oder Acylamino sind;
    jede Instanz von R2, R3, R4, R5, R8, R9, R10 und R11 unabhängig Wasserstoff, Hydroxyl, Halogen, Nitro, Alkyl, Alkenyl, Alkinyl, Cyan, Carboxyl, Sulfat, Amino, Alkoxy, Alkylamino, Alkylthio, Ether, Thioether, Ester, Amid, Thioester, Carbonat, Carbamat, Harnstoff, Sulfonat, Sulfon, Sulfoxid, Sulfonamid, Acyl, Acyloxy, Trialkylsilyloxy, Acylamino, Aryl, Heteroaryl, Carbocyclyl, Heterocyclylaralkyl, Aralkyloxy, Hetaralkyl, Carbocyclylalkyl oder Heterocyclylalkyl ist;
    R6 Wasserstoff, Hydroxyl, Halogen, Nitro, Cyan, Carboxyl, Sulfat, Alkyl, Alkenyl, Alkinyl, Amino, Alkoxy, Alkylamino, Alkylthio, Ether, Thioether, Ester, Amid, Thioester, Carbonat, Carbamat, Harnstoff, Sulfonat, Sulfon, Sulfoxid, Sulfonamid, Acyl, Acyloxy, Trialkylsilyloxy oder Acylamino ist; oder
    beliebige zwei von R1, R2, R3, R4, R5 und R6 zusammen mit den Kohlenstoffatomen, an denen sie angebracht sind, ein Aryl, Heteroaryl, Carbocyclyl oder Heterocyclyl bilden; oder
    beliebige zwei von R7, R8, R9, R10 und R11 zusammen mit den Kohlenstoffatomen, an denen sie angebracht sind, ein Aryl, Heteroaryl, Carbocyclyl oder Heterocyclyl bilden;
    und
    R12 H, Alkyl oder Aralkyl ist.
  2. Verfahren gemäß Anspruch 1, wobei der Übergangsmetallkatalysator einen Iridiumkomplex beinhaltet, wobei der Iridiumkatalysator optional durch das Vereinigen einer Iridiumquelle und eines chiralen Liganden hergestellt wird,
    wobei die Iridiumquelle ausgewählt ist aus (Acetylacetonato)(1,5-cyclooctadien)iridium(I), (Acetylacetonato)(1,5-cyclooctadien)iridium(I), (Acetylacetonato)dicarbonyliridium(I), Bis[1,2-bis(diphenylphosphino)ethan]carbonylchloriridium(I), Bis(1,5-cyclooctadien)diiridium(I)dichlorid, Bis(1,5-cyclooctadien)iridium(I)tetrafluorborat, Bis(cyclooctadien)iridium(I)tetrakis(3,5-bis(trifluormethyl)phenyl)borat, Chlorbis(cycloocten)iridium(I)dimer, (1,5-Cyclooctadien)bis(methyldiphenylphosphin)iridium(I)hexafluorphosphat, (1,5-Cyclooctadien)(hexafluoracetylacetonato)iridium(I), (1,5-Cyclooctadien)-η5-indenyl)iridium(I), (1,5-Cyclooctadien)(methoxy)iridium(I)dimer, (1,5-Cyclooctadien)(pyridin)(tricyclohexylphosphin)-iridium(I)hexafluorophosphat, (1,5-Cyclooctadien)(pyridin)(tricyclohexylphosphin)-iridium(I)hexafluorophosphat und (1,5-Cyclooctadien)(pyridin)(tricyclohexylphosphin)iridium(I)tetrakis[3,5-bis(trifluormethyl)phenyl]borat; und
    wobei der chirale Ligand einen Diphosphinliganden beinhaltet, der optional ausgewählt ist aus S-(CF3)-t-BuPHOX, S,S-Et-FerroTANE, S,Rp-xyliphos oder S,Rp-BTFM-xyliphos, R-(CF3)-t-BuPHOX, R,R-Et-FerroTANE, R,Sp-xyliphos und R,Sp -BTFM-xyliphos.
  3. Verfahren gemäß einem der vorhergehenden Ansprüche, wobei der Übergangsmetallkatalysator ein chiraler Übergangsmetallkatalysator ist und in einer Menge von etwa 0,1 Mol-% bis etwa 100 Mol-%, relativ zu der Verbindung der Formel (II), oder in einer Menge von etwa 5 Mol-% bis etwa 30 Mol-%, relativ zu der Verbindung der Formel (II), oder in einer Menge von etwa 20 Mol-%, relativ zu der Verbindung der Formel (II), verwendet wird.
  4. Verfahren gemäß einem der vorhergehenden Ansprüche, wobei die Verbindung der Formel (I) etwa 70 % ee oder mehr, etwa 80 % ee oder mehr, etwa 85 % ee oder mehr, etwa 88 % ee oder mehr, etwa 90 % ee oder mehr, etwa 91 % ee oder mehr, etwa 92 % ee oder mehr, etwa 93 % ee oder mehr, etwa 94 % ee oder mehr, etwa 95 % ee oder mehr, etwa 96 % ee oder mehr, etwa 97 % ee oder mehr, etwa 98 % ee oder mehr oder etwa 99 % ee oder mehr aufweist.
  5. Verfahren gemäß Anspruch 1, wobei das Herstellen der Verbindung der Formel (II):
    Figure imgb0083
    Folgendes beinhaltet: Vereinigen einer Verbindung der Formel (III):
    Figure imgb0084
    einer Verbindung der Formel (IV):
    Figure imgb0085
    und eines Übergangsmetallkatalysators unter Kreuzkopplungsbedingungen, wobei, solange Valenz und Stabilität es zulassen:
    R1 und R7 jeweils unabhängig Wasserstoff, Hydroxyl, Halogen, Nitro, Alkyl, Alkenyl, Alkinyl, Cyan, Carboxyl, Sulfat, Amino, Alkoxy, Alkylamino, Alkylthio, Ether, Thioether, Ester, Amid, Thioester, Carbonat, Carbamat, Harnstoff, Sulfonat, Sulfon, Sulfoxid, Sulfonamid, Acyl, Acyloxy, Alkylsilyloxy oder Acylamino sind;
    jede Instanz von R2, R3, R4, R5, R8, R9, R10 und R11 unabhängig Wasserstoff, Hydroxyl, Halogen, Nitro, Alkyl, Alkenyl, Alkinyl, Cyan, Carboxyl, Sulfat, Amino, Alkoxy, Alkylamino, Alkylthio, Ether, Thioether, Ester, Amid, Thioester, Carbonat, Carbamat, Harnstoff, Sulfonat, Sulfon, Sulfoxid, Sulfonamid, Acyl, Acyloxy, Alkylsilyloxy, Acylamino, Aryl, Heteroaryl, Carbocyclyl oder Heterocyclyl ist;
    R6 Wasserstoff, Hydroxyl, Halogen, Nitro, Alkyl, Alkenyl, Alkinyl, Cyan, Carboxyl, Sulfat, Amino, Alkoxy, Alkylamino, Alkylthio, Ether, Thioether, Ester, Amid, Thioester, Carbonat, Carbamat, Harnstoff, Sulfonat, Sulfon, Sulfoxid, Sulfonamid, Acyl, Acyloxy, Alkylsilyloxy oder Acylamino ist; oder
    beliebige zwei von R1, R2, R3, R4, R5 und R6 zusammen mit den Kohlenstoffatomen, an denen sie angebracht sind, ein Aryl, Heteroaryl, Carbocyclyl oder Heterocyclyl bilden; oder
    beliebige zwei von R7, R8, R9, R10 und R11 zusammen mit den Kohlenstoffatomen, an denen sie angebracht sind, ein Aryl, Heteroaryl, Carbocyclyl oder Heterocyclyl bilden; und
    R13 und R14 jeweils unabhängig Hydroxyl, Nitro, Cyan, Carboxyl, Sulfat, Amino, Alkoxy, Alkylamino, Alkylthio, Ether, Thioether, Ester, Amid, Thioester, Carbonat, Carbamat, Harnstoff, Sulfonat, Sulfon, Sulfoxid, Sulfonamid, Acyl, Acyloxy, Alkylsilyloxy oder Acylamino sind;
    wobei R13 und R14 optional jeweils unabhängig nichtsubstituiertes Alkyl oder mit einem oder mehreren Substituenten, ausgewählt aus Hydroxy, Alkoxy, Acyloxy, Amino und Thio, substituiertes Alkyl sind.
  6. Verfahren gemäß Anspruch 5, wobei der Übergangsmetallkatalysator einen Katalysator aus Nickel (z. B. Ni(COD)2), Palladium oder Platin beinhaltet; wobei der Übergangsmetallkatalysator optional einen Palladiumkatalysator beinhaltet, ausgewählt aus Pd/C, Pd2(DBA)3, Pd(PPh3)4, Pd(OC(O)RC)2, Pd(OAc)2, PdCl2, Pd(PhCN)2Cl2, Pd(CH3CN)2Cl2, PdBr2, Pd(acac)2, [Pd(allyl)CI]2, Pd(TFA)2, Pd2(pmdba)3, Pd(P(t-Bu)2Me)2 und vorgebildeten Pd(II)-Ligandenkomplexen;
    wobei RC optional substituiertes Alkyl, Alkenyl, Alkinyl, Aryl, Heteroaryl, Aralkyl, Heteroaralkyl, Cycloalkyl, Heterocycloalkyl, (Cycloalkyl)alkyl oder (Heterocycloalkyl)alkyl ist; und
    wobei optional der Übergangsmetallkatalysator in einer Menge von etwa 0,5 Mol-% bis etwa 50 Mol-%, relativ zu der Verbindung von Formel (III) oder (IV), verwendet wird.
  7. Verfahren gemäß einem der vorhergehenden Ansprüche, wobei R1 Wasserstoff ist;
    oder R1 nichtsubstituiertes Alkyl oder mit einem oder mehreren Substituenten, ausgewählt aus Hydroxy, Alkoxy, Acyloxy, Amino und Thio, substituiertes Alkyl ist; und/oder
    R2 aus H, Halogen und Hydroxyl ausgewählt ist; und/oder
    R3 Alkyl ist; und/oder
    R4 Alkoxy ist; und/oder
    R5 Hydroxy oder Alkoxy ist; und/oder
    R6 nichtsubstituiertes Alkyl oder mit einem oder mehreren Substituenten, ausgewählt aus Hydroxy, Alkoxy, Acyloxy, Amino und Thio, substituiertes Alkyl ist, R6 optional Hydroxyalkyl oder Acyloxyalkyl ist, R6 zum Beispiel CH2OH ist.
  8. Verfahren gemäß Anspruch 1, wobei R7 nichtsubstituiertes Alkyl oder mit einem oder mehreren Substituenten, ausgewählt aus Hydroxy, Alkoxy, Acyloxy, Amino und Thio, substituiertes Alkyl ist oder R7 H, Halogen oder Hydroxyl ist; und/oder
    R8 Alkoxy ist; und/oder
    R9 Alkyl ist; und/oder
    R10 Hydroxy oder Alkoxy ist; und/oder
    R11 H ist.
  9. Verfahren gemäß Anspruch 1 oder 6, wobei jede Instanz von R2, R3, R4, R5, R8, R9, R10 und R11 unabhängig nichtsubstituiertes Alkyl oder mit einem oder mehreren Substituenten, ausgewählt aus Hydroxy, Alkoxy, Acyloxy, Amino und Thio, substituiertes Alkyl ist.
EP18834872.6A 2017-07-19 2018-07-18 Verfahren zur herstellung von verbindungen mit bis-tetrahydroisochinolin Active EP3655379B1 (de)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201762534493P 2017-07-19 2017-07-19
PCT/US2018/042710 WO2019018539A1 (en) 2017-07-19 2018-07-18 PROCESSES FOR PREPARING COMPOUNDS CONTAINING BIS-TETRAHYDROISOQUINOLINE

Publications (3)

Publication Number Publication Date
EP3655379A1 EP3655379A1 (de) 2020-05-27
EP3655379A4 EP3655379A4 (de) 2021-03-24
EP3655379B1 true EP3655379B1 (de) 2023-04-05

Family

ID=65016370

Family Applications (1)

Application Number Title Priority Date Filing Date
EP18834872.6A Active EP3655379B1 (de) 2017-07-19 2018-07-18 Verfahren zur herstellung von verbindungen mit bis-tetrahydroisochinolin

Country Status (3)

Country Link
US (2) US10526334B2 (de)
EP (1) EP3655379B1 (de)
WO (1) WO2019018539A1 (de)

Families Citing this family (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP3655379B1 (de) * 2017-07-19 2023-04-05 California Institute of Technology Verfahren zur herstellung von verbindungen mit bis-tetrahydroisochinolin
CN110669006A (zh) * 2019-10-22 2020-01-10 中国科学技术大学 茚并异喹啉类化合物及其制备方法
CN114010639A (zh) * 2021-11-01 2022-02-08 济宁医学院 (-)-jorunnamycin A在抑制膀胱肿瘤细胞生长中的应用及应用方法

Family Cites Families (19)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4172896A (en) 1978-06-05 1979-10-30 Dainippon Pharmaceutical Co., Ltd. Methane-sulfonamide derivatives, the preparation thereof and composition comprising the same
GB9217295D0 (en) 1992-08-14 1992-09-30 Wellcome Found Controlled released tablets
US5541231A (en) 1993-07-30 1996-07-30 Glaxo Wellcome Inc. Stabilized Pharmaceutical
GB9315856D0 (en) 1993-07-30 1993-09-15 Wellcome Found Stabilized pharmaceutical
US5358970A (en) 1993-08-12 1994-10-25 Burroughs Wellcome Co. Pharmaceutical composition containing bupropion hydrochloride and a stabilizer
SG42925A1 (en) 1993-10-01 1997-10-17 Ciba Geigy Ag Fluoroalkyl-substituted ferrocenyl diphosphines as ligands for homogeneous catalysts
CN1229110C (zh) 1997-07-29 2005-11-30 阿尔康实验室公司 含半乳甘露聚糖聚合物和硼酸盐的眼用组合物
JP2002501892A (ja) 1998-01-29 2002-01-22 セプラコア インコーポレーテッド 光学的に純粋な(−)−ビュープロピオンの薬学的使用
US8889112B2 (en) 1999-09-16 2014-11-18 Ocularis Pharma, Llc Ophthalmic formulations including selective alpha 1 antagonists
EP1254140A4 (de) 2000-01-19 2003-03-12 Univ Columbia Verbindungen der saframycin-ecteinascidin-serie, verwendungen und synthesen davon
CA2447553A1 (en) 2000-11-03 2002-05-23 President And Fellows Of Harvard College Saframycins, analogues and uses thereof
EP1360337A1 (de) * 2001-02-09 2003-11-12 The Regents Of The University Of California Mitglieder der ecteinascidin familie: zusammensetzungen und methoden
US6933289B2 (en) 2003-07-01 2005-08-23 Allergan, Inc. Inhibition of irritating side effects associated with use of a topical ophthalmic medication
BRPI0413381A (pt) 2003-08-07 2006-10-17 Allergan Inc composições para liberação de compostos terapêuticos nos olhos e processos para fabricação e uso das mesmas
US20050059744A1 (en) 2003-09-12 2005-03-17 Allergan, Inc. Methods and compositions for the treatment of pain and other alpha 2 adrenergic-mediated conditions
WO2006066183A2 (en) 2004-12-16 2006-06-22 Axys Pharmaceuticals, Inc. Novel saframycin analogs as therapeutic agents
GB0522082D0 (en) 2005-10-31 2005-12-07 Pharma Mar Sa Formulations
JP2009046452A (ja) 2007-08-22 2009-03-05 Erick M Carreira ホスホルアミダイト配位子およびそれを用いたアリルアミンの製造方法
EP3655379B1 (de) * 2017-07-19 2023-04-05 California Institute of Technology Verfahren zur herstellung von verbindungen mit bis-tetrahydroisochinolin

Also Published As

Publication number Publication date
WO2019018539A1 (en) 2019-01-24
US10526334B2 (en) 2020-01-07
EP3655379A4 (de) 2021-03-24
US20200157102A1 (en) 2020-05-21
EP3655379A1 (de) 2020-05-27
US10781213B2 (en) 2020-09-22
US20190048009A1 (en) 2019-02-14

Similar Documents

Publication Publication Date Title
AU2020286278B2 (en) Compositions and methods for inhibiting arginase activity
US10781213B2 (en) Methods for preparing bis-tetrahydroisoquinoline-containing compounds
ES2934181T3 (es) Inhibidores de la interacción MDM2-P53, basados en isoindolinona, con actividad antineoplásica
TWI723047B (zh) 抗真菌化合物方法
EP2611778B1 (de) Guanidinverbindungen und zusammensetzungen zur hemmung von nampt
TW202035371A (zh) 抑制shp2活性化合物之製造方法、及由酸加成產生之產物
JP7027393B2 (ja) ベンズアミド及びニコチンアミド化合物及びこれを使用する方法
JP2019518726A (ja) アルギナーゼ阻害剤およびそれらの治療適用
KR102653190B1 (ko) 고 활성 sting 단백질 작용제 화합물
CA2653664C (en) Phosphine transition metal complex, process for producing same, and anticancer agent
JP2020515546A (ja) Mdm2−p53相互作用のイソインドリノン阻害剤およびそれを作製するためのプロセス
TW201904942A (zh) 經取代5-氰基吲哚化合物及其用途
US20210009547A1 (en) Substituted chromanes, analogs thereof, and methods of use and synthesis
UA100867C2 (uk) Похідна (аза)індолу, заміщена в положенні 5, фармацевтична композиція, що містить її, проміжні сполуки та спосіб їх одержання
EA029744B1 (ru) Ингибиторы киназ на основе простых ариловых эфиров
EP1610783A2 (de) Modulierung des zelltodes
KR102599437B1 (ko) Ror 감마 조정제로서의 트리시클릭 술폰
EP3753926A1 (de) Spiroverbindung als indolamin-2,3-dioxygenase-inhibitor
JP2022552693A (ja) Pd-l1拮抗薬化合物
KR102033727B1 (ko) 1-아미노인단 및 이의 제조방법
EP3805212A1 (de) 3-oxazolinonverbindungen, verfahren zu ihrer herstellung und ihre pharmazeutische anwendung
US20160101177A1 (en) pH AND LIGHT ACTIVATED ANTI-CANCER DRUGS
JP6067553B2 (ja) レニウム錯体及びその薬学的使用
JPH11506111A (ja) スワインソニンの新規誘導体、それらの調製方法及び治療薬としてのそれらの使用
CN102816129B (zh) 异噻唑啉(烷)酮取代的苯二羧酸衍生物及其作为β-分泌酶抑制剂的用途

Legal Events

Date Code Title Description
STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE INTERNATIONAL PUBLICATION HAS BEEN MADE

PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: REQUEST FOR EXAMINATION WAS MADE

17P Request for examination filed

Effective date: 20200219

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

AX Request for extension of the european patent

Extension state: BA ME

DAV Request for validation of the european patent (deleted)
DAX Request for extension of the european patent (deleted)
REG Reference to a national code

Ref country code: DE

Ref legal event code: R079

Ref document number: 602018048156

Country of ref document: DE

Free format text: PREVIOUS MAIN CLASS: C07B0043040000

Ipc: C07D0471180000

A4 Supplementary search report drawn up and despatched

Effective date: 20210224

RIC1 Information provided on ipc code assigned before grant

Ipc: A61K 31/4995 20060101ALI20210217BHEP

Ipc: C07D 471/18 20060101AFI20210217BHEP

Ipc: C07B 43/04 20060101ALI20210217BHEP

Ipc: A61P 35/00 20060101ALI20210217BHEP

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: EXAMINATION IS IN PROGRESS

17Q First examination report despatched

Effective date: 20220218

GRAP Despatch of communication of intention to grant a patent

Free format text: ORIGINAL CODE: EPIDOSNIGR1

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: GRANT OF PATENT IS INTENDED

INTG Intention to grant announced

Effective date: 20221031

GRAS Grant fee paid

Free format text: ORIGINAL CODE: EPIDOSNIGR3

GRAA (expected) grant

Free format text: ORIGINAL CODE: 0009210

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE PATENT HAS BEEN GRANTED

AK Designated contracting states

Kind code of ref document: B1

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

REG Reference to a national code

Ref country code: GB

Ref legal event code: FG4D

REG Reference to a national code

Ref country code: DE

Ref legal event code: R096

Ref document number: 602018048156

Country of ref document: DE

REG Reference to a national code

Ref country code: CH

Ref legal event code: EP

REG Reference to a national code

Ref country code: AT

Ref legal event code: REF

Ref document number: 1558191

Country of ref document: AT

Kind code of ref document: T

Effective date: 20230415

REG Reference to a national code

Ref country code: IE

Ref legal event code: FG4D

REG Reference to a national code

Ref country code: LT

Ref legal event code: MG9D

REG Reference to a national code

Ref country code: NL

Ref legal event code: MP

Effective date: 20230405

REG Reference to a national code

Ref country code: AT

Ref legal event code: MK05

Ref document number: 1558191

Country of ref document: AT

Kind code of ref document: T

Effective date: 20230405

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: NL

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20230405

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: SE

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20230405

Ref country code: PT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20230807

Ref country code: NO

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20230705

Ref country code: ES

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20230405

Ref country code: AT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20230405

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: GB

Payment date: 20230731

Year of fee payment: 6

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: RS

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20230405

Ref country code: PL

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20230405

Ref country code: LV

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20230405

Ref country code: LT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20230405

Ref country code: IS

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20230805

Ref country code: HR

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20230405

Ref country code: GR

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20230706

Ref country code: AL

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20230405

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: FR

Payment date: 20230725

Year of fee payment: 6

Ref country code: DE

Payment date: 20230728

Year of fee payment: 6

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: FI

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20230405

REG Reference to a national code

Ref country code: DE

Ref legal event code: R097

Ref document number: 602018048156

Country of ref document: DE

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: SK

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20230405

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: SM

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20230405

Ref country code: SK

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20230405

Ref country code: RO

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20230405

Ref country code: EE

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20230405

Ref country code: DK

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20230405

Ref country code: CZ

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20230405

PLBE No opposition filed within time limit

Free format text: ORIGINAL CODE: 0009261

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: NO OPPOSITION FILED WITHIN TIME LIMIT

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: MC

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20230405

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: MC

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20230405

REG Reference to a national code

Ref country code: CH

Ref legal event code: PL

26N No opposition filed

Effective date: 20240108

REG Reference to a national code

Ref country code: BE

Ref legal event code: MM

Effective date: 20230731

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: LU

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20230718

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: LU

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20230718

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: CH

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20230731

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: SI

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20230405